Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Int Immunopharmacol ; 136: 112278, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38815353

RESUMO

Intestinal stem cells (ISCs) are pivotal for the maintenance and regeneration of the intestinal epithelium. Berberine (BBR) exhibits diverse biological activities, but it remains unclear whether BBR can modulate ISCs' function. Therefore, we investigated the effects of BBR on ISCs in healthy and radiation-injured mice and explored the potential underlying mechanisms involved. The results showed that BBR significantly increased the length of the small intestines, the height of the villi, and the depth and density of the crypts, promoted the proliferation of cryptal epithelial cells and increased the number of OLFM4+ ISCs and goblet cells. Crypts from the BBR-treated mice were more capable of growing into enteroids than those from untreated mice. BBR alleviated WAI-induced intestinal injury. BBR suppressed the apoptosis of crypt epithelial cells, increased the quantity of goblet cells, and increased the quantity of OLFM4+ ISCs and tdTomato+ progenies of ISCs after 8 Gy WAI-induced injury. Mechanistically, BBR treatment caused a significant increase in the quantity of p-S6, p-STAT3 and p-ERK1/2 positive cryptal epithelial cells under physiological conditions and after WAI-induced injury. In conclusion, BBR is capable of enhancing the function of ISCs either physiologically or after radiation-induced injury, indicating that BBR has potential value in the treatment of radiation-induced intestinal injury.


Assuntos
Berberina , Mucosa Intestinal , Camundongos Endogâmicos C57BL , Células-Tronco , Animais , Berberina/farmacologia , Berberina/uso terapêutico , Células-Tronco/efeitos dos fármacos , Camundongos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos da radiação , Mucosa Intestinal/patologia , Masculino , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Lesões Experimentais por Radiação/tratamento farmacológico , Lesões Experimentais por Radiação/patologia , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/efeitos da radiação , Células Caliciformes/patologia , Lesões por Radiação/tratamento farmacológico , Lesões por Radiação/patologia , Fator de Transcrição STAT3/metabolismo , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/efeitos da radiação , Intestino Delgado/patologia , Intestino Delgado/lesões , Intestinos/efeitos dos fármacos , Intestinos/efeitos da radiação
2.
Int Immunopharmacol ; 131: 111908, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38518594

RESUMO

Radiation exposure often leads to serious health problems in humans. The intestinal epithelium is sensitive to radiation damage, and radiation causes destruction of the intestinal epithelial barrier, which leads to radiation enteritis (RE), the loss of fluids, and the translocation of intestinal bacteria and toxins; radiation can even threaten survival. In this study, we aimed to explore the influence of IVIg on the integrity of the intestinal epithelial barrier after RE. Using a RE mouse model, we investigated the protective effects of intravenous immunoglobulin (IVIg) on the epithelial junctions of RE mice and validated these findings with intestinal organoids cultured in vitro. In addition, transmission electron microscopy (TEM), western blotting (WB) and immunostaining were used to further investigate changes in intestinal epithelial ferroptosis and related signaling pathways. When RE occurs, the intestinal epithelial barrier is severely damaged. IVIg treatment significantly ameliorated this damage to epithelial tight junctions both in vivo and in vitro. Notably, IVIg alleviated RE by inhibiting intestinal epithelial ferroptosis in RE mice. Mechanistically, IVIg promoted activation of the mTOR pathway and inhibited ferroptosis in the intestinal epithelium of mice. Rapamycin, which is a potent inhibitor of the mTOR protein, significantly abolished the protective effect of IVIg against radiation-induced damage to intestinal epithelial tight junctions. Overall, IVIg can prevent RE-induced damage to the intestinal epithelial barrier and inhibit ferroptosis by activating the mTOR pathway; this study provides a new treatment strategy for patients with RE caused by radiotherapy or accidental nuclear exposure.


Assuntos
Enterite , Ferroptose , Exposição à Radiação , Humanos , Camundongos , Animais , Imunoglobulinas Intravenosas/farmacologia , Imunoglobulinas Intravenosas/uso terapêutico , Intestinos , Mucosa Intestinal , Serina-Treonina Quinases TOR/metabolismo
3.
Sci Adv ; 7(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33523835

RESUMO

Acute myeloid leukemia (AML) is a deadly hematological malignancy with frequent disease relapse. The biggest challenge for AML therapy is the lack of methods to target and kill the heterogeneous leukemia cells, which lead to disease relapse. Here, we describe a near-infrared (NIR) fluorescent dye, IR-26, which preferentially accumulates in the mitochondria of AML cells, depending on the hyperactive glycolysis of malignant cell, and simultaneously impairs oxidative phosphorylation (OXPHOS) to exert targeted therapeutic effects for AML cells. In particular, IR-26 also exhibits potential for real-time monitoring of AML cells with an in vivo flow cytometry (IVFC) system. Therefore, IR-26 represents a novel all-in-one agent for the integration of AML targeting, detection, and therapy, which may help to monitor disease progression and treatment responses, prevent unnecessary delays in administering upfront therapy, and improve therapeutic efficiency to the residual AML cells, which are responsible for disease relapse.


Assuntos
Antineoplásicos , Leucemia Mieloide Aguda , Antineoplásicos/farmacologia , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Recidiva
4.
Br J Pharmacol ; 177(17): 3991-4006, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32472692

RESUMO

BACKGROUND AND PURPOSE: There is an urgent but unmet need for mitigating radiation-induced intestinal toxicity while radio sensitising tumours for abdominal radiotherapy. We aimed to investigate the effects of metformin on radiation-induced intestinal toxicity and radiosensitivity of colorectal tumours. EXPERIMENTAL APPROACH: Acute and chronic histological injuries of the intestine from mice were used to assess radioprotection and IEC-6 cell line was used to investigate the mechanisms in vitro. The fractionated abdominal radiation model of HCT116 and HT29 tumour grafts was used to determine the effects on colorectal cancer. KEY RESULTS: Metformin alleviated radiation-induced acute and chronic intestinal toxicity by optimising mitophagy which was AMPK-dependent. In addition, our data indicated that metformin increased the radiosensitivity of colorectal tumours with P53 mutation both in vitro and in vivo. CONCLUSION AND IMPLICATIONS: Metformin may be a radiotherapy adjuvant agent for colorectal cancers especially those carrying P53 mutation. Our findings provide a new strategy for further precise clinical trials for metformin on radiotherapy.


Assuntos
Neoplasias Colorretais , Metformina , Animais , Apoptose , Autofagia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/radioterapia , Metformina/farmacologia , Camundongos , Mutação , Proteína Supressora de Tumor p53/genética
5.
Nat Commun ; 10(1): 2538, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31182708

RESUMO

The pathological mechanisms of radiation ulcer remain unsolved and there is currently no effective medicine. Here, we demonstrate that persistent DNA damage foci and cell senescence are involved in radiation ulcer development. Further more, we identify cordycepin, a natural nucleoside analogue, as a potent drug to block radiation ulcer (skin, intestine, tongue) in rats/mice by preventing cell senescence through the increase of NRF2 nuclear expression (the assay used is mainly on skin). Finally, cordycepin is also revealed to activate AMPK by binding with the α1 and γ1 subunit near the autoinhibitory domain of AMPK, then promotes p62-dependent autophagic degradation of Keap1, to induce NRF2 dissociate from Keap1 and translocate to the nucleus. Taken together, our findings identify cordycepin prevents radiation ulcer by inhibiting cell senescence via NRF2 and AMPK in rodents, and activation of AMPK or NRF2 may thus represent therapeutic targets for preventing cell senescence and radiation ulcer.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Senescência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Desoxiadenosinas/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Lesões Experimentais por Radiação/prevenção & controle , Úlcera/prevenção & controle , Animais , Apoptose , Linhagem Celular , Senescência Celular/efeitos da radiação , Dano ao DNA/efeitos da radiação , Desoxiadenosinas/toxicidade , Fibroblastos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Lesões Experimentais por Radiação/tratamento farmacológico , Lesões Experimentais por Radiação/patologia , Ratos Sprague-Dawley , Úlcera/tratamento farmacológico , Úlcera/patologia , Raios X/efeitos adversos
6.
Adv Mater ; : e1800475, 2018 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-29961960

RESUMO

The endoplasmic reticulum (ER) stress signaling or unfolded protein response (UPR) is a common feature of many human diseases, including cancer. Excessive activation of ER stress directly induces cell death, holding a new promising strategy for the therapeutic intervention of cancer. Current ER-stress-inducing agents mainly target UPR components or proteasomes, which exert limited treatment efficacy and undesired side effects due to unselective ER stress and poor tumor-specific distribution. In this study, a unique near-infrared (NIR) fluorophore, IR-34, is synthesized and identified to selectively and efficiently trigger tumoricidal ER stress by targeting the mitochondrial protein NDUFS1. IR-34 is demonstrated to specifically accumulate in living cancer cells for tumor NIR imaging and drastically inhibit tumor growth and recurrence without causing apparent toxicity. Thus, this multifunctional NIR fluorophore may represent a novel theranostic agent for tumor imaging-guided treatment and also strengthens the idea that mitochondria could be a useful target for therapeutic ER stress in cancer cells.

7.
Adv Sci (Weinh) ; 5(3): 1700392, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29593950

RESUMO

The characterization of cancer stem-like cells (CSCs) has profound implications for elucidating cancer biology and developing treatment strategies. Although surface markers are already used to identify CSCs, the expression of these markers is controversially linked to the phenotypes in different types of tumors and does not represent all functionally relevant of CSCs. Very recently, hyperactive HIF-1α/glycolysis metabolic pathway is recognized as a master regulator of CSCs. In this study, a near-infrared fluorescent small-molecule, IR-780, is identified for the exclusive characterization of human CSCs through the HIF-1α/glycolysis dependent mitochondrial transporter ABCB10's activity. The results identified for the first time that ABCB10 is involved in the preferential uptake of IR-780 in CSCs, which is regulated by HIF-1α via the direct interaction with the binding site of ABCB10 gene promoter region. In addition, IR-780 is demonstrated to conjugate with anticancer drug 5-fluorouracil to act as a potential drug delivery carrier for CSC-targeted therapy. Thus, the studies provide a new rational approach independent of surface markers to characterize CSCs via small-molecule-based imaging of HIF-1α/glycolysis hyperactive metabolic pathway dependent mitochondrial transporter's activity, which holds promise for the further development of CSCs targeted diagnostic and therapeutic strategies.

8.
EMBO J ; 37(7)2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29467218

RESUMO

In 1900, Adami speculated that a sequence of context-independent energetic and structural changes governed the reversion of differentiated cells to a proliferative, regenerative state. Accordingly, we show here that differentiated cells in diverse organs become proliferative via a shared program. Metaplasia-inducing injury caused both gastric chief and pancreatic acinar cells to decrease mTORC1 activity and massively upregulate lysosomes/autophagosomes; then increase damage associated metaplastic genes such as Sox9; and finally reactivate mTORC1 and re-enter the cell cycle. Blocking mTORC1 permitted autophagy and metaplastic gene induction but blocked cell cycle re-entry at S-phase. In kidney and liver regeneration and in human gastric metaplasia, mTORC1 also correlated with proliferation. In lysosome-defective Gnptab-/- mice, both metaplasia-associated gene expression changes and mTORC1-mediated proliferation were deficient in pancreas and stomach. Our findings indicate differentiated cells become proliferative using a sequential program with intervening checkpoints: (i) differentiated cell structure degradation; (ii) metaplasia- or progenitor-associated gene induction; (iii) cell cycle re-entry. We propose this program, which we term "paligenosis", is a fundamental process, like apoptosis, available to differentiated cells to fuel regeneration following injury.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Regeneração/fisiologia , Células Acinares , Animais , Autofagossomos/fisiologia , Ciclo Celular/fisiologia , Transdiferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Celulas Principais Gástricas/patologia , Trato Gastrointestinal/patologia , Expressão Gênica , Humanos , Lisossomos , Metaplasia/genética , Camundongos , Camundongos Endogâmicos C57BL , Fase S/fisiologia , Fatores de Transcrição SOX9/metabolismo , Estômago/lesões , Estômago/patologia , Transferases (Outros Grupos de Fosfato Substituídos)/genética
9.
Artigo em Inglês | MEDLINE | ID: mdl-29158275

RESUMO

The increasing incidence of multidrug-resistant Acinetobacter baumannii (MDRAb) infections worldwide has necessitated the development of novel antibiotics. Human defensin 5 (HD5) is an endogenous peptide with a complex architecture and antibacterial activity against MDRAb In the present study, we attempted to simplify the structure of HD5 by removing disulfide bonds. We found that the Cys2-4 bond was most indispensable for HD5 to inactivate MDRAb, although the antibacterial activity of the derivative was significantly attenuated. We then replaced the noncationic and nonhydrophobic residues with electropositive Arg to increase the antibacterial activity of HD5 derivative that contains a Cys2-4 bond, obtaining another derivative termed HD5d5. The in vitro antibacterial assay and irradiation-wound-infection animal experiment both showed that HD5d5 was much more effective than HD5 at eliminating MDRAb Further investigations revealed that HD5d5 efficiently bound to outer membrane lipid A and penetrated membranes, leading to bacterial collapse and peptide translocation. Compared to HD5, more HD5d5 molecules were located in the cytoplasm of MDRAb, and HD5d5 was more efficient at reducing the activities of superoxide dismutase and catalase, causing the accumulation of reactive oxygen species that are detrimental to microbes. In addition, HD5 failed to suppress the pathogenic outer membrane protein A of Acinetobacter baumannii (AbOmpA) at concentrations up to 50 µg/ml, whereas HD5d5 strongly bound to AbOmpA and exhibited a dramatic toxin-neutralizing ability, thus expanding the repertoire of drugs that is available to treat MDRAb infections.


Assuntos
Infecções por Acinetobacter/tratamento farmacológico , Acinetobacter baumannii/efeitos dos fármacos , Antibacterianos/farmacologia , Regulação Bacteriana da Expressão Gênica , Infecção dos Ferimentos/tratamento farmacológico , alfa-Defensinas/farmacologia , Infecções por Acinetobacter/microbiologia , Infecções por Acinetobacter/mortalidade , Infecções por Acinetobacter/patologia , Acinetobacter baumannii/genética , Acinetobacter baumannii/crescimento & desenvolvimento , Acinetobacter baumannii/metabolismo , Animais , Antibacterianos/síntese química , Proteínas da Membrana Bacteriana Externa/antagonistas & inibidores , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Catalase/antagonistas & inibidores , Catalase/genética , Catalase/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Humanos , Lipídeo A/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ligação Proteica , Engenharia de Proteínas/métodos , Isoformas de Proteínas/síntese química , Isoformas de Proteínas/farmacologia , Transporte Proteico , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/antagonistas & inibidores , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Análise de Sobrevida , Irradiação Corporal Total , Infecção dos Ferimentos/microbiologia , Infecção dos Ferimentos/mortalidade , Infecção dos Ferimentos/patologia , alfa-Defensinas/síntese química
10.
Genes Dev ; 31(2): 154-171, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-28174210

RESUMO

We hypothesized that basic helix-loop-helix (bHLH) MIST1 (BHLHA15) is a "scaling factor" that universally establishes secretory morphology in cells that perform regulated secretion. Here, we show that targeted deletion of MIST1 caused dismantling of the secretory apparatus of diverse exocrine cells. Parietal cells (PCs), whose function is to pump acid into the stomach, normally lack MIST1 and do not perform regulated secretion. Forced expression of MIST1 in PCs caused them to expand their apical cytoplasm, rearrange mitochondrial/lysosome trafficking, and generate large secretory granules. Mist1 induced a cohort of genes regulated by MIST1 in multiple organs but did not affect PC function. MIST1 bound CATATG/CAGCTG E boxes in the first intron of genes that regulate autophagosome/lysosomal degradation, mitochondrial trafficking, and amino acid metabolism. Similar alterations in cell architecture and gene expression were also caused by ectopically inducing MIST1 in vivo in hepatocytes. Thus, MIST1 is a scaling factor necessary and sufficient by itself to induce and maintain secretory cell architecture. Our results indicate that, whereas mature cell types in each organ may have unique developmental origins, cells performing similar physiological functions throughout the body share similar transcription factor-mediated architectural "blueprints."


Assuntos
Regulação da Expressão Gênica/genética , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Células Parietais Gástricas/citologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Via Secretória/genética , Células Acinares/citologia , Células Acinares/efeitos dos fármacos , Células Acinares/metabolismo , Animais , Antineoplásicos Hormonais/farmacologia , Linhagem Celular , Expressão Ectópica do Gene/efeitos dos fármacos , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/ultraestrutura , Tamoxifeno/farmacologia
11.
Gastroenterology ; 152(4): 762-766.e7, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27932312

RESUMO

Parietal cell atrophy is considered to cause metaplasia in the stomach. We developed mice that express the diphtheria toxin receptor specifically in parietal cells to induce their death, and found this to increase proliferation in the normal stem cell zone and neck but not to cause metaplastic reprogramming of chief cells. Furthermore, the metaplasia-inducing agents tamoxifen or DMP-777 still induced metaplasia even after previous destruction of parietal cells by diphtheria toxin. Atrophy of parietal cells alone therefore is not sufficient to induce metaplasia: completion of metaplastic reprogramming of chief cells requires mechanisms beyond parietal cell injury or death.


Assuntos
Apoptose , Celulas Principais Gástricas/patologia , Células Parietais Gástricas/patologia , Células Parietais Gástricas/fisiologia , Estômago/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Atrofia/induzido quimicamente , Azetidinas , Proliferação de Células , Reprogramação Celular , Celulas Principais Gástricas/metabolismo , Toxina Diftérica/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Peptídeos e Proteínas de Sinalização Intercelular , Fator Intrínseco/metabolismo , Metaplasia/induzido quimicamente , Metaplasia/genética , Metaplasia/metabolismo , Camundongos , Células Parietais Gástricas/efeitos dos fármacos , Peptídeos/metabolismo , Piperazinas , Lectinas de Plantas/metabolismo , Tamoxifeno
12.
Sci Rep ; 6: 25314, 2016 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-27126764

RESUMO

Progressive liver disease is a major health issue for which no effective treatment is available, leading to cirrhosis and orthotopic liver transplantation. However, the lack of availability of donor organs and other adverse factors including rejection limit its extensive clinical application. Cell-based therapy using mesenchymal stem/stromal cells (MSCs) may represent an attractive therapeutic option. Dermal-derived mesenchymal cells (DMCs) are attractive as one of the abundant sources from which to isolate mesenchymal cells for therapeutic applications and can be easily accessed with minimal harm to the donor. In this study, we used two different animal models to investigate potential therapeutic effect of DMCs transplantation in liver injury. We found that DMCs administration alleviated liver fibrosis and restored the liver function in fibrotic mice induced by CCl4. Furthermore, in an acute irradiation induced damage model, a unique population of DMCs could engraft into the liver tissue for a long period, exhibiting the phenotype of both mesenchymal cells and macrophage cells, and improve the survival of mice exposed to 8 Gy lethally total-body irradiation. These discoveries provide important evidence that DMCs therapy has a beneficial effect on liver injury, and provide new insight into liver injury therapy depending on the alternative cells.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/terapia , Regeneração Hepática , Transplante de Células-Tronco Mesenquimais , Lesões Experimentais por Radiação/terapia , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Modelos Animais de Doenças , Cirrose Hepática/patologia , Cirrose Hepática/terapia , Testes de Função Hepática , Camundongos , Lesões Experimentais por Radiação/patologia , Pele/citologia , Resultado do Tratamento
13.
Mol Cell Biochem ; 378(1-2): 107-16, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23456479

RESUMO

Although mesenchymal stem cells (MSCs) promote lung cancer growth in vivo, in vitro studies indicate that they inhibit the proliferation of lung cancer cells. Because malignant tumors contain a heterogeneous cell population with variable capacity for self-renewal, the aim of this study was to determine whether the inconsistencies between in vitro and in vivo studies are a result of differential effects of MSCs on the heterogeneous cell population within lung cancer cell lines. Human MSCs were isolated from the bone marrow, and their cell surface antigen expression and multi-lineage differentiation capacity was examined at passage 10. CD133+ cells were isolated from A549 and H446 cell lines using immunomagnetic separation. The effects of MSCs on the growth and microsphere formation of heterogeneous cell populations within two lung cancer cell lines (A549 and H446) were compared. MSCs inhibited the in vitro proliferation of both cell lines, but significantly accelerated tumor formation and stimulated tumor growth in vivo (P < 0.05). In CD133+ cells isolated from both A549 and H446 cells, co-culture with MSCs for 1-3 days significantly increased their proliferation (P < 0.05). MSCs also significantly increased microsphere formation in both cell lines (P < 0.05). Selective stimulation of CD133+ cell growth may account for the discrepant effects of MSCs on lung cancer progression.


Assuntos
Neoplasias Pulmonares/patologia , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Neoplásicas/metabolismo , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Feminino , Glicoproteínas/metabolismo , Humanos , Separação Imunomagnética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Peptídeos/metabolismo
14.
J Surg Res ; 183(1): 427-34, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23462453

RESUMO

BACKGROUND: Bone marrow-derived mesenchymal stem cells (BM-MSCs) play a crucial role in tissue repair. Their role in thermal burn wound regeneration and the relevant mechanism, however, is rarely studied. METHODS: BM-MSCs from green fluorescent protein transgenic male mice were transfused to irradiated recipient female C57BL/6 mice. Twenty-one days later, the female mice were inflicted with burn wounds. The size of the burned area was measured by an in vivo fluorescence imaging system, and BM-MSC chemotaxis and epithelialization were estimated by fluorescence in situ hybridization and immunofluorescence technology. The expression of CXCL12 and CXCR4 in the wound margin was detected by enzyme-linked immunosorbent assay and immunohistochemistry. The importance of CXCL12/CXCR4 signaling in BM-MSC chemotaxis was further estimated by blocking CXCR4 in vivo and in vitro. RESULTS: In vivo imaging results showed that BM-MSCs migrated to the injured margins. Fluorescence in situ hybridization and immunofluorescence technology revealed that Y chromosome-positive cells derived from green fluorescent protein transgenic mice were detected to be colocalized with keratin protein. Enzyme-linked immunosorbent assay revealed increased levels of CXCL12 and CXCR4 protein in the wound sites of BM-MSC-treated chimeric mice after burn. Immunohistochemistry also disclosed that CXCL12 levels were elevated at postburn day 7 compared with day 0. Furthermore, pretreatment of the BM-MSCs with the CXCR4 antagonist AMD3100 significantly inhibited the mobilization of BM-MSCs in vitro and in vivo, which attenuated wound closure. CONCLUSION: BM-MSC migration to the burned margins promotes the epithelialization of the wound, and mobilization of BM-MSCs is mediated by CXCL12/CXCR4 signaling.


Assuntos
Queimaduras/metabolismo , Quimiocina CXCL12/metabolismo , Células-Tronco Mesenquimais/fisiologia , Reepitelização , Receptores CXCR4/metabolismo , Animais , Quimiotaxia , Quimera , Células Epidérmicas , Feminino , Proteínas de Fluorescência Verde , Folículo Piloso/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
15.
Tumour Biol ; 34(1): 145-54, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23055190

RESUMO

Bone marrow-derived stem cells (BMDCs) have the ability to differentiate into lung epithelial cells in response to damage; however, their role in squamous cell carcinoma (SCC) formation is unknown. This study aimed to determine whether BMDC-derived lung epithelial cells could contribute to SCC formation. A model of lung SCC induced with N-nitrosodiethylamine (NDEA) in recipient female mice transplanted with green fluorescent protein (GFP)-positive BMDCs from male donors was established. Incorporation of BMDCs in lung tissue was determined using immunohistochemistry and immunofluorescence to detect GFP expression and fluorescence in situ hybridization to Y chromosomes. BMDC appeared at three stages of lung SCC progression: metaplasia, dysplasia, and carcinoma. There was a significantly higher proportion of GFP-positive (GFP(+)) cells within SCC than was found in metaplasia and dysplasia 16 weeks post-transplantation (both P < 0.017); GFP(+) BMDCs were also observed in clusters within several SCC nests. Furthermore, most GFP(+) cells in SCC were pancytokeratin-positive (PCK(+)) epithelial cells, and some exhibited proliferative activity as determined by Ki67 staining (9.7 ± 3.92 %). The presence of GFP(+)Ki67(+)PCK(+) cells within SCC nests suggested that some donor BMDCs differentiated into proliferating epithelial cells. Finally, analysis of p63 expression, a marker of SCC cells, indicated that the presence of GFP(+)p63(+) cells (green) in inner parts of the SCC. These findings strongly suggest that BMDC-derived lung epithelial cells could participate in lung SCC formation and partially contribute to tumor growth, which might have significant potential implications for both clinical cancer therapy using BMDCs.


Assuntos
Células da Medula Óssea/fisiologia , Carcinoma de Células Escamosas/patologia , Células Epiteliais/fisiologia , Neoplasias Pulmonares/patologia , Células-Tronco/fisiologia , Animais , Transplante de Medula Óssea , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/metabolismo , Diferenciação Celular , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica , Dietilnitrosamina , Feminino , Proteínas de Fluorescência Verde , Queratinócitos/patologia , Queratinas/biossíntese , Antígeno Ki-67/análise , Neoplasias Pulmonares/induzido quimicamente , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfoproteínas/análise , Transativadores/análise
16.
Mol Cell Biochem ; 364(1-2): 29-37, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22198336

RESUMO

Steroid receptor coactivator-3 (SRC-3) is a multifunctional protein that plays an important role in mammary gland growth, development, and tumorigenesis. In this study, SCR-3 gene knockout mice were used to study the effects of SCR-3 on the immunosuppression accompanied with systemic inflammatory response syndrome (SIRS). Bacterial clearance assay was performed by blood culture and frozen sections, and the results showed that the absence of SCR-3 protein serious damaged the innate immune system and the body's ability to inactivate or phagocytosis of bacteria was significantly decreased, and the absence of SCR-3 protein also weakened phagocytes' ability to degrade bacteria and their metabolites. Furthermore, animal model of inflammatory reaction was established and the immune function was determined, and the results revealed that SRC-3 protein may play an important role in maintenance of T-cells' immune function, and severe T-cell immune function disorder would be resulted once SRC-3 protein is missing. In addition, the results of our study showed the steady-state of lymphocyte subsets was destroyed after SIRS, leading the suppression of cellular immune function, and the absence of SCR-3 protein may aggravate the suppression of T-lymphocyte function. Therefore, the present study demonstrated that the absence of SCR-3 protein would aggravate immunosuppression. In addition, SRC-3 protein is a significant regulator of infection and inflammation, and SRC-3 protein play an essential role in the development of immunosuppression accompanied with SIRS.


Assuntos
Imunidade Inata , Coativador 3 de Receptor Nuclear/imunologia , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Linfócitos T/imunologia , Animais , Bactérias/patogenicidade , Feminino , Imunidade Inata/genética , Interleucina-2/sangue , Lipopolissacarídeos/administração & dosagem , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Knockout , Coativador 3 de Receptor Nuclear/deficiência , Coativador 3 de Receptor Nuclear/genética , Fagocitose/genética , Síndrome de Resposta Inflamatória Sistêmica/sangue , Síndrome de Resposta Inflamatória Sistêmica/genética , Linfócitos T/patologia
17.
Cell Transplant ; 20(5): 741-51, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21054944

RESUMO

Near-infrared (NIR) fluorescent agents hold great promise for noninvasive in vivo imaging. We have recently reported that a NIR fluorescent heptamethine dye, IR-780 iodide, exhibits unique optical properties for biomedical imaging. On the basis of this foregoing work, we further describe here the potential application of IR-780 iodide as a novel NIR agent for stem cell labeling and tracking. The labeling efficiency, subcellular localization, and the effects on cell viability and differentiation of IR-780 iodide were investigated. The in vivo distribution of stem cells after intravenous transplantation was traced by whole-body animal NIR imaging. Our results showed that IR-780 iodide exhibited superior labeling efficiency and biocompatibility with unique optical properties. Following whole-body NIR imaging, the pulmonary passage of stem cells was noninvasively visualized in rats after systemic transplantation of IR-780 iodide-labeled stem cells through intravenous delivery. With this NIR imaging method, we further confirmed that pretreatment with sodium nitroprusside (SNP), a vasodilator agent, significantly reduced the cell trapping in the lung and increased the cell passage through the lung capillaries. Our study suggests that IR-780 iodide may represent an effective NIR fluorophore for stem cell labeling and tracking.


Assuntos
Corantes Fluorescentes/química , Indóis/química , Espectroscopia de Luz Próxima ao Infravermelho , Células-Tronco/citologia , Animais , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Nitroprussiato/química , Ratos , Ratos Sprague-Dawley , Transplante de Células-Tronco , Células-Tronco/química , Imagem Corporal Total
18.
J Radiat Res ; 51(5): 527-33, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20921821

RESUMO

Vascular endothelial cells are very sensitive to ionizing radiation, and it is important to develop effective prevent agents and measures in radiation exposure protection. In the present study, the protective effects of atorvastatin on irradiated human umbilical vein endothelial cells (HUVEC) and the possible mechanisms were explored. Cultured HUVEC were treated by atorvastatin at a final concentration of 10 µ mol/ml for 10 minutes, and then irradiated at a dose of 2 Gy or 25 Gy. Twenty-four hours after irradiation, apoptosis of HUVEC was monitored by flow cytometry, and the expression of thrombomodulin (TM) and protein C activation in HUVEC was respectively assessed by flow cytometry and spectrophotometry. After treatment with atorvastatin for 24 h, the rate of cell apoptosis decreased by 6% and 16% in cells irradiated with 2 Gy and 25 Gy, respectively. TM expression increased by 77%, 59%, and 61% in untreated cells, 2 Gy irradiation-treated cells, and 25 Gy irradiation-treated cells, respectively. The protein C levels in 2 Gy and 25 Gy irradiation-treated cells were reduced by 23% and 34% when compared with untreated cells, but up-regulated by 79% and 76% when compared with cells which were irradiated and treated with atorvastatin. In conclusion, these data indicate that atorvastatin exerts protective effects on irradiated HUVEC by reducing apoptosis by up-regulating TM expression and enhancing protein C activation in irradiated HUVEC.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/efeitos da radiação , Ácidos Heptanoicos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Pirróis/farmacologia , Atorvastatina , Células Cultivadas , Humanos , Lesões por Radiação/tratamento farmacológico
19.
Biomaterials ; 31(36): 9535-43, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20875681

RESUMO

Failure to cure many cancers once they are disseminated has been attributed to the presence of resistant cancer stem cells. Cantharidin, a natural compound isolated from the beetles and other insects has been traditionally used as anticancer agent, but limited by its significant toxicity. It has shown that cantharidin can force cancer cells prematurely into cell cycle and subsequently induce apoptotic cell death through the inhibition of protein phosphatase 2A (PP2A). In this study, we showed that a synthesized analog of cantharidin, LB1, with significant PP2A inhibition activity but without apparent toxicity, greatly enhanced the effectiveness of the standard anti-sarcoma chemotherapeutic agent, doxorubicin (DOX), in the xenograft growth inhibition and lung metastases prevention of an aggressive sarcoma derived from transformed mesenchymal stem cells in syngeneic rats. We report here on the possibility of, pharmacologic inhibition of PP2A with low toxicity cantharidin derivatives may be a useful strategy to enhance the effectiveness of DNA-damaged chemotherapeutic drugs against stem cell-derived cancer.


Assuntos
Cantaridina/análogos & derivados , Cantaridina/uso terapêutico , Transformação Celular Neoplásica/patologia , Doxorrubicina/uso terapêutico , Células-Tronco Mesenquimais/patologia , Sarcoma/tratamento farmacológico , Sarcoma/patologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores/metabolismo , Cantaridina/farmacologia , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Doxorrubicina/farmacologia , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Multipotentes/metabolismo , Metástase Neoplásica , Ratos , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Biomed Biotechnol ; 2010: 164986, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20396598

RESUMO

Interstitial cells of Cajal (ICCs) in gastrointestinal tract are specialized cells serving as pacemaker cells. The origin of ICCs is currently not fully characterized. In this work, we aimed to study whether bone marrow-derived cells (BMDCs) could contribute to the origin of ICCs in the muscular plexus of small intestine using GFP-C57BL/6 chimeric mice.Engraftment of BMDCs in the intestine was investigated for GFP expression. GFP positive bone marrow mononuclear cells reached a proportion of 95.65% +/- 3.72% at different times in chimerism. Donor-derived cells distributed widely in all the layers of the gastrointestinal tract. There were GFP positive BMDCs in the myenteric plexus, which resembled characteristics of ICCs, including myenteric location, c-Kit positive staining, and ramified morphology. Donor-derived ICCs in the myenteric plexus contributed to a percentage ranging 9.25% +/- 4.9% of all the ICCs in the myenteric plexus. In conclusion, here we described that donor-derived BMDCs might differentiate into gastrointestinal ICCs after radiation injury, which provided an alternative source for the origin of the ICCs in the muscular plexus of adult intestine. These results further identified the plasticity of BMDCs and indicated therapeutic implications of BMDCs for the gastrointestinal dysmotility caused by ICCs disorders.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/fisiologia , Células Intersticiais de Cajal/patologia , Lesões Experimentais por Radiação/patologia , Animais , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Células Intersticiais de Cajal/efeitos da radiação , Intestino Delgado/patologia , Intestino Delgado/efeitos da radiação , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA